Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 113(8): 926-937, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28444132

ABSTRACT

AIMS: Endothelial nitric oxide (NO) synthase (eNOS) is known to play a cardioprotective protective. However, the molecular mechanisms regulating eNOS activity during ischaemia/reperfusion (I/R) injury are incompletely understood. eNOS is a substrate for several kinases that positively or negatively affect its enzymatic activity. Herein, we sought to correlate eNOS phosphorylation status with cardiomyocyte survival and we investigated the contribution of the proline-rich tyrosine kinase 2 (PYK2)/eNOS axis to the regulation of myocardial infarct size in vivo. METHODS AND RESULTS: Exposure of H9c2 cardiomyocytes to H2O2 lead to PYK2 phosphorylation on its activator site (Y402) and eNOS phosphorylation on the inhibitor site Y656 and the activator site S1176. Both H2O2-induced eNOS phosphorylation events were abolished by PYK2 pharmacological inhibition or gene knockdown. Activity assays demonstrated that phosphorylation of the tyrosine inhibitory site exerts a dominant effect over S1176. In cardiomyocytes subjected to oxidative stress or oxygen-glucose deprivation, inhibition of PYK2 limited cell injury; this effect was prevented by inhibition of NO production. In vivo, ischaemia-reperfusion induced an early activation of PYK2, leading to eNOS phosphorylation on Y656, which, in turn, reduced NO output, as judged by the low tissue levels of its downstream effector cGMP. Moreover, pharmacological blockade of PYK2 alleviated eNOS inhibition and prevented cardiac damage following I/R injury in wild-type, but not in eNOS KO mice. CONCLUSION: The current studies demonstrate that PYK2 is a pivotal regulator of eNOS function in myocardial infarction and identify PYK2 as a novel therapeutic target for cardioprotection.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Myocardium/metabolism , Nitric Oxide Synthase Type III/metabolism , Tyrosine/metabolism , Animals , Enzyme Activation/drug effects , Hydrogen Peroxide/pharmacology , Male , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Phosphorylation , Rats
2.
Pharmacol Res ; 111: 556-562, 2016 09.
Article in English | MEDLINE | ID: mdl-27378567

ABSTRACT

Soluble guanylate cyclase (sGC) is a receptor for nitric oxide (NO). Binding of NO to ferrous (Fe(2+)) heme increases its catalytic activity, leading to the production of cGMP from GTP. Hydrogen sulfide (H2S) is a signaling molecule that exerts both direct and indirect anti-oxidant effects. In the present, study we aimed to determine whether H2S could regulate sGC redox state and affect its responsiveness to NO-releasing agents and sGC activators. Using cultured rat aortic smooth muscle cells, we observed that treatment with H2S augmented the response to the NO donor DEA/NO, while attenuating the response to the heme-independent activator BAY58-2667 that targets oxidized sGC. Similarly, overexpression of H2S-synthesizing enzyme cystathionine-γ lyase reduced the ability of BAY58-2667 to promote cGMP accumulation. In experiments with phenylephrine-constricted mouse aortic rings, treatment with rotenone (a compound that increases ROS production), caused a rightward shift of the DEA/NO concentration-response curve, an effect partially restored by H2S. When rings were pre-treated with H2S, the concentration-response curve to BAY 58-2667 shifted to the right. Using purified recombinant human sGC, we observed that treatment with H2S converted ferric to ferrous sGC enhancing NO-donor-stimulated sGC activity and reducing BAY 58-2667-triggered cGMP formation. The present study identified an additional mechanism of cross-talk between the NO and H2S pathways at the level of redox regulation of sGC. Our results provide evidence that H2S reduces sGC heme Fe, thus, facilitating NO-mediated cellular signaling events.


Subject(s)
Heme/metabolism , Hydrogen Sulfide/pharmacology , Nitric Oxide/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Benzoates/pharmacology , Cells, Cultured , Cystathionine gamma-Lyase/metabolism , In Vitro Techniques , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Donors/pharmacology , Oxidation-Reduction , Phenylephrine , Quaternary Ammonium Compounds/pharmacology , Rats
3.
Handb Exp Pharmacol ; 230: 337-63, 2015.
Article in English | MEDLINE | ID: mdl-26162843

ABSTRACT

Hydrogen sulfide is rapidly emerging as a key physiological mediator and potential therapeutic tool in numerous areas such as acute and chronic inflammation, neurodegenerative and cardiovascular disease, diabetes, obesity and cancer. However, the vast majority of the published studies have employed crude sulfide salts such as sodium hydrosulfide (NaSH) and sodium sulfide (Na2S) as H2S "donors" to generate H2S. Although these salts are cheap, readily available and easy to use, H2S generated from them occurs as an instantaneous and pH-dependent dissociation, whereas endogenous H2S synthesis from the enzymes cystathionine γ-lyase, cystathionine-ß-synthase and 3-mercaptopyruvate sulfurtransferase is a slow and sustained process. Furthermore, sulfide salts are frequently used at concentrations (e.g. 100 µM to 10 mM) far in excess of the levels of H2S reported in vivo (nM to low µM). For the therapeutic potential of H2S is to be properly harnessed, pharmacological agents which generate H2S in a physiological manner and deliver physiologically relevant concentrations are needed. The phosphorodithioate GYY4137 has been proposed as "slow-release" H2S donors and has shown promising efficacy in cellular and animal model diseases such as hypertension, sepsis, atherosclerosis, neonatal lung injury and cancer. However, H2S generation from GYY4137 is inefficient necessitating its use at high concentrations/doses. However, structural modification of the phosphorodithioate core has led to compounds (e.g. AP67 and AP105) with accelerated rates of H2S generation and enhanced biological activity. In this review, the therapeutic potential and limitations of GYY4137 and related phosphorodithioate derivatives are discussed.


Subject(s)
Hydrogen Sulfide/metabolism , Morpholines/pharmacology , Organothiophosphorus Compounds/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Cytoprotection , Humans , Morpholines/therapeutic use , Organothiophosphorus Compounds/therapeutic use
4.
Am J Physiol Lung Cell Mol Physiol ; 308(7): L683-92, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25595645

ABSTRACT

Inspiratory resistive breathing (IRB) is characterized by large negative intrathoracic pressures and was shown to induce pulmonary inflammation in previously healthy rats. Matrix metalloproteinases (MMP)-9 and -12 are induced by inflammation and mechanical stress in the lung. We hypothesized that IRB induces MMP-9 and -12 in the lung. Anesthetized, tracheostomized rats breathed spontaneously through a two-way valve, connected to an inspiratory resistance, with the tidal inspiratory tracheal pressure set at 50% of the maximum. Quietly breathing animals served as controls. After 3 and 6 h of IRB, respiratory mechanics were measured, bronchoalveolar lavage (BAL) was performed, lung injury score was estimated, and lung MMP-9 was estimated by zymography and ELISA. MMP-9 and MMP-12 immunohistochemistry was performed. Isolated normal alveolar macrophages were incubated with BAL from rats that underwent IRB. After 18 h, MMP-9 and -12 levels were measured in supernatants, and immunocytochemistry was performed. Macrophages were treated with IL-1ß, IL-6, or TNF-α, and MMP-9 in supernatants was measured. After 6 h of IRB, leukocytes in BAL increased, and IL-1ß and IL-6 levels were elevated. Elasticity and injury score were increased after 3 and 6 h of IRB. Lung MMP-9 levels increased after 6 h of IRB. MMP-9 and MMP-12 were detected in alveolar macrophages and epithelial (bronchial/alveolar) cells after 3 and 6 h of IRB. MMP-9 and MMP-12 were found in supernatants after treatment with 6 h of IRB BAL. Cytosolic immunostaining was detected after treatment with 3 and 6 h of IRB BAL. All cytokines induced MMP-9 in culture supernatants. In conclusion, IRB induces MMP-9 and -12 in the lung of previously healthy rats.


Subject(s)
Dyspnea/enzymology , Lung/enzymology , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Cells, Cultured , Enzyme Induction , Female , Macrophages, Alveolar/enzymology , Protein Transport , Rats, Wistar , Respiration
5.
Nitric Oxide ; 46: 7-13, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25553675

ABSTRACT

The importance of hydrogen sulfide (H2S) in physiology and disease is being increasingly recognized in recent years. Unlike nitric oxide (NO) that signals mainly through soluble guanyl cyclase (sGC)/cGMP, H2S is more promiscuous, affecting multiple pathways. It interacts with ion channels, enzymes, transcription factors and receptors. It was originally reported that H2S does not alter the levels of cyclic nucleotides. More recent publications, however, have shown increases in intracellular cGMP following exposure of cells or tissues to exogenously administered or endogenously produced H2S. Herein, we discuss the evidence for the participation of cGMP in H2S signaling and reconcile the seemingly divergent results presented in the literature on the role of this cyclic nucleotide in the biological actions of H2S.


Subject(s)
Cyclic GMP/metabolism , Hydrogen Sulfide/metabolism , Signal Transduction , Animals , Cyclic GMP/chemistry , Humans , Hydrogen Sulfide/chemistry , Mice
6.
Am J Respir Cell Mol Biol ; 52(6): 762-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25353067

ABSTRACT

Inspiratory resistive breathing (RB), encountered in obstructive lung diseases, induces lung injury. The soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP) pathway is down-regulated in chronic and acute animal models of RB, such as asthma, chronic obstructive pulmonary disease, and in endotoxin-induced acute lung injury. Our objectives were to: (1) characterize the effects of increased concurrent inspiratory and expiratory resistance in mice via tracheal banding; and (2) investigate the contribution of the sGC/cGMP pathway in RB-induced lung injury. Anesthetized C57BL/6 mice underwent RB achieved by restricting tracheal surface area to 50% (tracheal banding). RB for 24 hours resulted in increased bronchoalveolar lavage fluid cellularity and protein content, marked leukocyte infiltration in the lungs, and perturbed respiratory mechanics (increased tissue resistance and elasticity, shifted static pressure-volume curve right and downwards, decreased static compliance), consistent with the presence of acute lung injury. RB down-regulated sGC expression in the lung. All manifestations of lung injury caused by RB were exacerbated by the administration of the sGC inhibitor, 1H-[1,2,4]oxodiazolo[4,3-]quinoxalin-l-one, or when RB was performed using sGCα1 knockout mice. Conversely, restoration of sGC signaling by prior administration of the sGC activator BAY 58-2667 (Bayer, Leverkusen, Germany) prevented RB-induced lung injury. Strikingly, direct pharmacological activation of sGC with BAY 58-2667 24 hours after RB reversed, within 6 hours, the established lung injury. These findings raise the possibility that pharmacological targeting of the sGC-cGMP axis could be used to ameliorate lung dysfunction in obstructive lung diseases.


Subject(s)
Guanylate Cyclase/metabolism , Lung Diseases, Obstructive/enzymology , Lung Injury/enzymology , Airway Resistance , Animals , Benzoates/pharmacology , Benzoates/therapeutic use , Cyclic GMP/metabolism , Drug Evaluation, Preclinical , Enzyme Activation , Guanylate Cyclase/antagonists & inhibitors , Lung Diseases, Obstructive/drug therapy , Lung Injury/drug therapy , Male , Mice, Inbred C57BL
7.
Cardiovasc Res ; 102(1): 138-47, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24501330

ABSTRACT

AIMS: Therapeutic use of sulfhydrylated inhibitor S-zofenopril has raised different hypotheses regarding the role played by its thiol group in the beneficial clinical effects exerted compared with other angiotensin-converting enzyme (ACE) inhibitors. Here, we investigated hydrogen sulfide (H2S) pathway as accountable for extra-beneficial effects in vascular function. METHODS AND RESULTS: Spontaneously hypertensive rat (SHRs) and control Wistar Kyoto (WKY) rats were treated with either S-zofenopril or enalapril in vivo. Aorta and carotid were harvested and ex vivo vascular reactivity to acetylcholine (Ach) and L-cysteine (L-cys) assessed. Cystathionine-ß-synthase (CBS), cystathionine-γ-lyase (CSE), and 3-mercaptosulfur-transferase (3MST) expression, as well as H2S levels, were evaluated in both vascular tissues. The vascular response to Ach in both carotid and aorta was impaired in SHR (~30%, P < 0.001). S-zofenopril, but not enalapril, restored this response, while L-cys-induced relaxation was enhanced. CSE expression in vessels and tissue/plasma H2S levels were restored to WKY values in SHRs receiving S-zofenopril. In contrast, CBS and 3MST expression were not modified by treatments. S-zofenoprilat, an active metabolite of S-zofenopril, releases H2S in a 'cell-free' assay and it directly relaxed vessels in vitro in a concentration-dependent manner (P < 0.001). In vivo administration of R-zofenoprilat diasteroisomer, which does not inhibit ACE, did not modify blood pressure; nonetheless, it retained the beneficial effect on SHR vascular function as well as restored plasma/tissue H2S levels. CONCLUSION: Our findings establish that S-zofenopril improves vascular function by potentiating the H2S pathway in a model of spontaneous hypertension. This novel mechanism, unrelated to ACE inhibition and based on H2S release, could explain the beneficial effects of sulfhydrylated ACE inhibitors reported in the clinical literature.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Aorta/drug effects , Blood Pressure/drug effects , Captopril/analogs & derivatives , Hydrogen Sulfide/pharmacology , Animals , Captopril/chemistry , Captopril/pharmacology , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Male , Rats, Inbred SHR , Rats, Inbred WKY
8.
J Med Chem ; 56(21): 8948-8952, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24090476

ABSTRACT

Recently, the structure of BAY 58-2667 bound to the Nostoc sp. H-NOX domain was published. On the basis of this structural information, we designed BAY 58-2667 derivatives and tested their effects on soluble guanylyl cyclase (sGC) activity. Derivative 20 activated sGC 4.8-fold more than BAY 58-2667. Co-crystallization of 20 with the Ns H-NOX domain revealed that the increased conformational distortion at the C-terminal region of αF helix containing 110-114 residues contributes to the higher activation triggered by 20.


Subject(s)
Benzoates/pharmacology , Guanylate Cyclase/metabolism , Heme/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Benzoates/chemical synthesis , Benzoates/chemistry , Dose-Response Relationship, Drug , Drug Design , Enzyme Activation , Heme/chemistry , Models, Molecular , Molecular Conformation/drug effects , Nitric Oxide/metabolism , Nostoc/enzymology , Receptors, Cytoplasmic and Nuclear/agonists , Soluble Guanylyl Cyclase , Structure-Activity Relationship
9.
Br J Pharmacol ; 169(4): 922-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23488457

ABSTRACT

BACKGROUND AND PURPOSE: Hydrogen sulfide (H2S) is a signalling molecule that belongs to the gasotransmitter family. Two major sources for endogenous enzymatic production of H2S are cystathionine ß synthase (CBS) and cystathionine γ lyase (CSE). In the present study, we examined the selectivity of commonly used pharmacological inhibitors of H2S biosynthesis towards CSE and CBS. EXPERIMENTAL APPROACH: To address this question, human CSE or CBS enzymes were expressed and purified from Escherichia coli as fusion proteins with GSH-S-transferase. After purification, the activity of the recombinant enzymes was tested using the methylene blue method. KEY RESULTS: ß-Cyanoalanine (BCA) was more potent in inhibiting CSE than propargylglycine (PAG) (IC50 14 ± 0.2 µM vs. 40 ± 8 µM respectively). Similar to PAG, L-aminoethoxyvinylglycine (AVG) only inhibited CSE, but did so at much lower concentrations. On the other hand, aminooxyacetic acid (AOAA), a frequently used CBS inhibitor, was more potent in inhibiting CSE compared with BCA and PAG (IC50 1.1 ± 0.1 µM); the IC50 for AOAA for inhibiting CBS was 8.5 ± 0.7 µM. In line with our biochemical observations, relaxation to L-cysteine was blocked by AOAA in aortic rings that lacked CBS expression. Trifluoroalanine and hydroxylamine, two compounds that have also been used to block H2S biosynthesis, blocked the activity of CBS and CSE. Trifluoroalanine had a fourfold lower IC50 for CBS versus CSE, while hydroxylamine was 60-fold more selective against CSE. CONCLUSIONS AND IMPLICATIONS: In conclusion, although PAG, AVG and BCA exhibit selectivity in inhibiting CSE versus CBS, no selective pharmacological CBS inhibitor is currently available.


Subject(s)
Cystathionine beta-Synthase/antagonists & inhibitors , Cystathionine gamma-Lyase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alkynes/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Hydrogen Sulfide/analysis , Hydrogen Sulfide/metabolism , In Vitro Techniques , Kinetics , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Vasodilation/drug effects
10.
Am J Physiol Heart Circ Physiol ; 303(5): H597-604, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22730391

ABSTRACT

Nitric oxide (NO) by activating soluble guanylyl cyclase (sGC) is involved in vascular homeostasis via induction of smooth muscle relaxation. In cardiovascular diseases (CVDs), endothelial dysfunction with altered vascular reactivity is mostly attributed to decreased NO bioavailability via oxidative stress. However, in several studies, relaxation to NO is only partially restored by exogenous NO donors, suggesting sGC impairment. Conflicting results have been reported regarding the nature of this impairment, ranging from decreased expression of one or both subunits of sGC to heme oxidation. We showed that sGC activity is impaired by thiol S-nitrosation. Recently, angiotensin II (ANG II) chronic treatment, which induces hypertension, was shown to generate nitrosative stress in addition to oxidative stress. We hypothesized that S-nitrosation of sGC occurs in ANG II-induced hypertension, thereby leading to desensitization of sGC to NO hence vascular dysfunction. As expected, ANG II infusion increases blood pressure, aorta remodeling, and protein S-nitrosation. Intravital microscopy indicated that cremaster arterioles are resistant to NO-induced vasodilation in vivo in anesthetized ANG II-treated rats. Concomitantly, NO-induced cGMP production decreases, which correlated with S-nitrosation of sGC in hypertensive rats. This study suggests that S-nitrosation of sGC by ANG II contributes to vascular dysfunction. This was confirmed in vitro by using A7r5 smooth muscle cells infected with adenoviruses expressing sGC or cysteine mutants: ANG II decreases NO-stimulated activity in the wild-type but not in one mutant, C516A. This result indicates that cysteine 516 of sGC mediates ANG II-induced desensitization to NO in cells.


Subject(s)
Angiotensin II , Guanylate Cyclase/metabolism , Hypertension/chemically induced , Muscle, Smooth, Vascular/enzymology , Nitric Oxide/metabolism , Oxidative Stress , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Arterioles/enzymology , Arterioles/physiopathology , Blood Pressure , Cell Line , Cyclic GMP/metabolism , Cysteine , Disease Models, Animal , Enzyme Activation , Guanylate Cyclase/genetics , Hypertension/enzymology , Hypertension/physiopathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Mutation , Myocytes, Smooth Muscle/enzymology , Nitric Oxide Donors/pharmacology , Nitrosation , Oxidative Stress/drug effects , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Soluble Guanylyl Cyclase , Time Factors , Transfection , Vascular Resistance , Vasodilation
11.
Bioorg Med Chem ; 20(8): 2675-8, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22436388

ABSTRACT

Thioglycine and l-thiovaline are stable under acidic and basic conditions but in the presence of bicarbonate they liberate the gasotransmitter H(2)S. In cells both thioamino acids were proven to enhance cGMP formation and promote vasorelaxation in mouse aortic rings. Given that H(2)S is known to lower arterial hypertension, reduce oxidative stress and exhibit cardioprotective effects in preclinical models, H(2)S donors hold promise as novel treatments for cardiovascular diseases.


Subject(s)
Amino Acids/pharmacology , Aorta/drug effects , Glycine/analogs & derivatives , Glycine/pharmacology , Hydrogen Sulfide/metabolism , Myocytes, Smooth Muscle/drug effects , Valine/analogs & derivatives , Valine/pharmacology , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Aorta/metabolism , Cyclic GMP/biosynthesis , Cyclic GMP/metabolism , Hydrogen Sulfide/chemistry , Mice , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Rats
12.
PLoS One ; 7(12): e53319, 2012.
Article in English | MEDLINE | ID: mdl-23285278

ABSTRACT

A growing body of evidence suggests that hydrogen sulfide (H2S) is a signaling molecule in mammalian cells. In the cardiovascular system, H2S enhances vasodilation and angiogenesis. H2S-induced vasodilation is hypothesized to occur through ATP-sensitive potassium channels (K(ATP)); however, we recently demonstrated that it also increases cGMP levels in tissues. Herein, we studied the involvement of cGMP-dependent protein kinase-I in H2S-induced vasorelaxation. The effect of H2S on vessel tone was studied in phenylephrine-contracted aortic rings with or without endothelium. cGMP levels were determined in cultured cells or isolated vessel by enzyme immunoassay. Pretreatment of aortic rings with sildenafil attenuated NaHS-induced relaxation, confirming previous findings that H2S is a phosphodiesterase inhibitor. In addition, vascular tissue levels of cGMP in cystathionine gamma lyase knockouts were lower than those in wild-type control mice. Treatment of aortic rings with NaHS, a fast releasing H2S donor, enhanced phosphorylation of vasodilator-stimulated phosphoprotein in a time-dependent manner, suggesting that cGMP-dependent protein kinase (PKG) is activated after exposure to H2S. Incubation of aortic rings with a PKG-I inhibitor (DT-2) attenuated NaHS-stimulated relaxation. Interestingly, vasodilatory responses to a slowly releasing H2S donor (GYY 4137) were unaffected by DT-2, suggesting that this donor dilates mouse aorta through PKG-independent pathways. Dilatory responses to NaHS and L-cysteine (a substrate for H2S production) were reduced in vessels of PKG-I knockout mice (PKG-I⁻/⁻). Moreover, glibenclamide inhibited NaHS-induced vasorelaxation in vessels from wild-type animals, but not PKG-I⁻/⁻, suggesting that there is a cross-talk between K(ATP) and PKG. Our results confirm the role of cGMP in the vascular responses to NaHS and demonstrate that genetic deletion of PKG-I attenuates NaHS and L-cysteine-stimulated vasodilation.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/physiology , Hydrogen Sulfide/pharmacology , Vasodilation/drug effects , Animals , Aorta/drug effects , Aorta/physiology , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Male , Mice , Mice, Knockout , Phosphodiesterase 5 Inhibitors/pharmacology , Rats , Rats, Wistar , Vasodilation/genetics
13.
Proc Natl Acad Sci U S A ; 108(33): 13829-34, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21808008

ABSTRACT

The goal of the present studies was to investigate the role of changes in hydrogen sulfide (H(2)S) homeostasis in the pathogenesis of hyperglycemic endothelial dysfunction. Exposure of bEnd3 microvascular endothelial cells to elevated extracellular glucose (in vitro "hyperglycemia") induced the mitochondrial formation of reactive oxygen species (ROS), which resulted in an increased consumption of endogenous and exogenous H(2)S. Replacement of H(2)S or overexpression of the H(2)S-producing enzyme cystathionine-γ-lyase (CSE) attenuated the hyperglycemia-induced enhancement of ROS formation, attenuated nuclear DNA injury, reduced the activation of the nuclear enzyme poly(ADP-ribose) polymerase, and improved cellular viability. In vitro hyperglycemia resulted in a switch from oxidative phosphorylation to glycolysis, an effect that was partially corrected by H(2)S supplementation. Exposure of isolated vascular rings to high glucose in vitro induced an impairment of endothelium-dependent relaxations, which was prevented by CSE overexpression or H(2)S supplementation. siRNA silencing of CSE exacerbated ROS production in hyperglycemic endothelial cells. Vascular rings from CSE(-/-) mice exhibited an accelerated impairment of endothelium-dependent relaxations in response to in vitro hyperglycemia, compared with wild-type controls. Streptozotocin-induced diabetes in rats resulted in a decrease in the circulating level of H(2)S; replacement of H(2)S protected from the development of endothelial dysfunction ex vivo. In conclusion, endogenously produced H(2)S protects against the development of hyperglycemia-induced endothelial dysfunction. We hypothesize that, in hyperglycemic endothelial cells, mitochondrial ROS production and increased H(2)S catabolism form a positive feed-forward cycle. H(2)S replacement protects against these alterations, resulting in reduced ROS formation, improved endothelial metabolic state, and maintenance of normal endothelial function.


Subject(s)
Endothelium, Vascular/drug effects , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hyperglycemia/drug therapy , Mitochondria/drug effects , Animals , Cell Line , Diabetes Mellitus, Experimental , Endothelial Cells , Glucose/pharmacology , Homeostasis , Hydrogen Sulfide/metabolism , Hyperglycemia/pathology , Mitochondria/metabolism , Protective Agents/therapeutic use , Rats , Reactive Oxygen Species/metabolism
14.
Vascul Pharmacol ; 53(5-6): 215-22, 2010.
Article in English | MEDLINE | ID: mdl-20813203

ABSTRACT

Vascular endothelial growth factor (VEGF) stimulates nitric oxide (NO) production, which mediates many of its angiogenic actions. However, the angiogenic pathways that operate downstream of NO following VEGF treatment are not well characterized. Herein, we used DT-2 and DT-3, two highly selective cGMP-dependent protein kinase I peptide inhibitors to determine the contribution of PKG-I in VEGF-stimulated angiogenesis. Incubation of chicken chorioallantoic membranes (CAM) with PKG-I peptide inhibitors decreased vascular length in a dose-dependent manner, with DT-3 being more effective than DT-2. Moreover, inhibition of PKG-I with DT-3 abolished the angiogenic response elicited by VEGF in the rabbit eye cornea. PKG-I inhibition also blocked VEGF-stimulated vascular leakage. In vitro, treatment of cells with VEGF stimulated phosphorylation of the PKG substrate VASP through VEGFR2 activation; the VEGF-stimulated VASP phosphorylation was reduced by DT-2. Pre-treatment of cells with DT-2 or DT-3 inhibited VEGF-stimulated mitogen-activated protein kinase cascades (ERK1/2 and p38), growth, migration and sprouting of endothelial cells. The above observations taken together identify PKG-I as a downstream effector of VEGFR2 in EC and provide a rational basis for the use of PKG-I inhibitors in disease states characterized by excessive neovascularization.


Subject(s)
Chorioallantoic Membrane/drug effects , Cornea/drug effects , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelial Cells/drug effects , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factors/pharmacology , Animals , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/blood supply , Cornea/blood supply , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelial Cells/metabolism , Fluoresceins/pharmacology , Humans , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Peptides/pharmacology , Phosphorylation , Rabbits , Signal Transduction , Vascular Endothelial Growth Factors/physiology
15.
Arterioscler Thromb Vasc Biol ; 30(10): 1998-2004, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20634473

ABSTRACT

OBJECTIVE: Recent studies have demonstrated that hydrogen sulfide (H(2)S) is produced within the vessel wall from L-cysteine regulating several aspects of vascular homeostasis. H(2)S generated from cystathione γ-lyase (CSE) contributes to vascular tone; however, the molecular mechanisms underlying the vasorelaxing effects of H(2)S are still under investigation. METHODS AND RESULTS: Using isolated aortic rings, we observed that addition of L-cysteine led to a concentration-dependent relaxation that was prevented by the CSE inhibitors DL-propargylglyicine (PAG) and ß-cyano-l-alanine (BCA). Moreover, incubation with PAG or BCA resulted in a rightward shift in sodium nitroprusside-and isoproterenol-induced relaxation. Aortic tissues exposed to PAG or BCA contained lower levels of cGMP, exposure of cells to exogenous H(2)S or overexpression of CSE raised cGMP concentration. RNA silencing of CSE expression reduced intracellular cGMP levels confirming a positive role for endogenous H(2)S on cGMP accumulation. The ability of H(2)S to enhance cGMP levels was greatly reduced by the nonselective phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine. Finally, addition of H(2)S to a cell-free system inhibited both cGMP and cAMP breakdown. CONCLUSIONS: These findings provide direct evidence that H(2)S acts as an endogenous inhibitor of phosphodiesterase activity and reinforce the notion that this gasotransmitter could be therapeutically exploited.


Subject(s)
Hydrogen Sulfide/metabolism , Phosphodiesterase Inhibitors/metabolism , Vasodilation/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alkynes/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Clone Cells , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Cysteine/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , In Vitro Techniques , Male , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Vasodilation/drug effects
16.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R824-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20032260

ABSTRACT

Nitric oxide (NO) is known to promote vascular endothelial growth factor (VEGF)-stimulated permeability and angiogenesis. However, effector molecules that operate downstream of NO in this pathway remain poorly characterized. Herein, we determined the effect of soluble guanylyl cyclase (sGC) inhibition on VEGF responses in vitro and in vivo. Treatment of endothelial cells (EC) with VEGF stimulated eNOS phosphorylation and cGMP accumulation; pretreatment with the sGC inhibitor 4H-8-bromo-1,2,4-oxadiazolo(3,4-d)benz(b)(1,4)oxazin-1-one (NS-2028) blunted cGMP levels without affecting VEGF-receptor phosphorylation. Incubation of cells with NS-2028 blocked the mitogenic effects of VEGF. In addition, cells in which sGC was inhibited exhibited no migration and sprouting in response to VEGF. To study the mechanisms through which NS-2028 inhibits EC migration, we determined the effects of alterations in cGMP levels on p38 MAPK. Initially, we observed that inhibition of sGC attenuated VEGF-stimulated activation of p38. In contrast, the addition of 8-Br-cGMP to EC stimulated p38 phosphorylation. The addition of cGMP elevating agents (BAY 41-2272, DETA NO and YC-1) enhanced EC migration. To test whether sGC also mediated the angiogenic effects of VEGF in vivo, we used the rabbit cornea assay. Animals receiving NS-2028 orally displayed a reduced angiogenic response to VEGF. As increased vascular permeability occurs prior to new blood vessel formation, we determined the effect of NS-2028 in vascular leakage. Using a modified Miles assay, we observed that NS-2028 attenuated VEGF-induced permeability. Overall, we provide evidence that sGC mediates the angiogenic and permeability-promoting activities of VEGF, indicating the significance of sGC as a downstream effector of VEGF-triggered responses.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Neovascularization, Physiologic/physiology , Oxadiazoles/pharmacology , Oxazines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Animals , Aorta/drug effects , Aorta/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Cornea/blood supply , Drug Interactions , Endothelial Cells/cytology , Fibroblast Growth Factor 2/pharmacology , Guanylate Cyclase/metabolism , Humans , Neovascularization, Physiologic/drug effects , Rabbits , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology , Vasodilation/drug effects , Vasodilation/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Interferon Cytokine Res ; 30(5): 321-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20035621

ABSTRACT

Strenuous exercise leads to the up-regulation of interleukin-6 (IL-6) production and enhanced nitric oxide (NO) release within the contracting skeletal muscles. In this study, we investigated whether NO regulates IL-6 production in C2C12 myotubes. These cells exhibited a concentration-dependent increase in IL-6 production upon stimulation with NO donors (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA-NONOate), (Z)-1-[N-(3-aminopropyl)-N-(n-propyl)amino]diazen-1-ium-1,2-diolate (PAPA-NONOate), and sodium nitroprusside (SNP). This treatment did not alter cGMP levels nor did the soluble guanylyl cyclase (sGC) inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one(ODQ), alter this response. The NO-independent sGC activator 5-cyclopropyl-2-[1-(2-fluoro-benzyl)-1H-pyrazolo[3,4-b]pyridin-3-yl]-pyrimidin-4-ylamine (BAY41-2272) and cyclic guanosine monophosphate (cGMP) analog 8Br-cGMP failed to induce IL-6 production. Upon exposure to NO donors, we observed an increase in Erk1/2 and p38 MAPK phosphorylation but not in SAPK/JNK. In addition, NO-induced IL-6 release was inhibited in a concentration-dependent fashion by the MEK1/2 inhibitor PD98059 and the p38 MAPK inhibitor SB203580 but not by the SAPK/JNK inhibitor SP600125. We conclude that NO-stimulated IL-6 production in differentiated C2C12 myotubes is cGMP-independent and mediated by activation of MAPK pathways.


Subject(s)
Cyclic GMP/metabolism , Interleukin-6/biosynthesis , Muscle Fibers, Skeletal/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Animals , Cell Line , Cyclic GMP/analogs & derivatives , Flavonoids/pharmacology , Imidazoles/pharmacology , Immunization , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Nitric Oxide/immunology , Oxadiazoles/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Quinoxalines/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
18.
Proc Natl Acad Sci U S A ; 106(51): 21972-7, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19955410

ABSTRACT

The goal of the current study was to investigate the role of exogenous and endogenous hydrogen sulfide (H(2)S) on neovascularization and wound healing in vitro and in vivo. Incubation of endothelial cells (ECs) with H(2)S enhanced their angiogenic potential, evidenced by accelerated cell growth, migration, and capillary morphogenesis on Matrigel. Treatment of chicken chorioallantoic membranes (CAMS) with H(2)S increased vascular length. Exposure of ECs to H(2)S resulted in increased phosphorylation of Akt, ERK, and p38. The K(ATP) channel blocker glibenclamide or the p38 inhibitor SB203580 abolished H(2)S-induced EC motility. Since glibenclamide inhibited H(2)S-triggered p38 phosphorylation, we propose that K(ATP) channels lay upstream of p38 in this process. When CAMs were treated with H(2)S biosynthesis inhibitors dl-propylargylglycine or beta-cyano-L-alanine, a reduction in vessel length and branching was observed, indicating that H(2)S serves as an endogenous stimulator of the angiogenic response. Stimulation of ECs with vascular endothelial growth factor (VEGF) increased H(2)S release, while pharmacological inhibition of H(2)S production or K(ATP) channels or silencing of cystathionine gamma-lyase (CSE) attenuated VEGF signaling and migration of ECs. These results implicate endothelial H(2)S synthesis in the pro-angiogenic action of VEGF. Aortic rings isolated from CSE knockout mice exhibited markedly reduced microvessel formation in response to VEGF when compared to wild-type littermates. Finally, in vivo, topical administration of H(2)S enhanced wound healing in a rat model, while wound healing was delayed in CSE(-/-) mice. We conclude that endogenous and exogenous H(2)S stimulates EC-related angiogenic properties through a K(ATP) channel/MAPK pathway.


Subject(s)
Endothelium, Vascular/drug effects , Hydrogen Sulfide/pharmacology , Neovascularization, Physiologic/drug effects , Apoptosis/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Signal Transduction/drug effects , Wound Healing/drug effects
19.
Am J Physiol Renal Physiol ; 296(6): F1386-95, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19297451

ABSTRACT

Chronic kidney disease (CKD) is associated with decreased renal nitric oxide (NO) production and increased plasma levels of methylarginines. The naturally occurring guanidino-methylated arginines N-monomethyl-l-arginine (l-NMMA) and asymmetric dimethyl-l-arginine (ADMA) inhibit NO synthase activity. We hypothesized that ADMA and l-NMMA compromise the integrity of the glomerular filtration barrier via NO depletion. We studied the effect of ADMA on albumin permeability (P(alb)) in isolated glomeruli and examined whether this effect involves NO- and superoxide (O(2)(*-))-dependent mechanisms. ADMA at concentrations found in circulation of patients with CKD decreased cGMP and increased P(alb) in a dose-dependent manner. A similar increase in P(alb) was caused by l-NMMA but at a concentration two orders of magnitude higher than that of ADMA. NO donor DETA-NONOate or cGMP analog abrogated the effect of ADMA on P(alb). The SOD mimetic tempol or the NAD(P)H oxidase inhibitor apocynin also prevented the ADMA-induced increase in P(alb). The NO-independent soluble guanylyl cyclase (sGC) activator BAY 41-2272, at concentrations that increased glomerular cGMP production, attenuated the ADMA-induced increase in P(alb). Furthermore, sGC incapacitation by the heme site-selective inhibitor ODQ increased P(alb). We conclude that ADMA compromises the integrity of the filtration barrier by altering the bioavailability of NO and O(2)(*-) and that NO-independent activation of sGC preserves the integrity of this barrier under conditions of NO depletion. NO-independent activation of sGS may be a useful pharmacotherapeutic approach for preservation of glomerular function in CKD thereby reducing the risk for cardiovascular events.


Subject(s)
Anthracenes/toxicity , Arginine/analogs & derivatives , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/metabolism , Nitric Oxide/metabolism , Propane/analogs & derivatives , Superoxides/metabolism , Albumins/metabolism , Animals , Arginine/pharmacology , Arginine/toxicity , Free Radical Scavengers/pharmacology , Glomerular Basement Membrane/pathology , Guanylate Cyclase/metabolism , Male , NADPH Oxidases/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Oxidation-Reduction , Permeability/drug effects , Propane/toxicity , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , omega-N-Methylarginine/toxicity
20.
J Natl Cancer Inst ; 100(20): 1464-76, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18840818

ABSTRACT

BACKGROUND: Tumor cells in malignant pleural effusions (MPEs) are an important source of monocyte chemoattractant protein (MCP)-1. However, the role of tumor-derived MCP-1 in the pathogenesis and progression of MPE has not been determined. METHODS: B16 mouse skin melanoma cells, which are deficient in MCP-1 expression, and mouse Lewis lung cancer (LLC) cells, which express high levels of MCP-1, were engineered to stably express MCP-1 and short hairpin RNAs (shRNAs) targeting the MCP-1 transcript, respectively. Cells were injected into the pleural cavities of syngeneic immunocompetent mice, and MPE volume and pleural tumors were quantified at necropsy (day 14). MCP-1 and other mediators were determined by cytometric bead array and enzyme-linked immunosorbent assay, and mononuclear and endothelial cells were identified by immunolabeling of F4/80 and factor VIII-related antigen respectively. Mouse survival was assessed using Kaplan-Meier analysis. Vascular permeability in mice with MPE was assessed using albumin-binding Evans blue. Statistical tests were two-sided. RESULTS: LLC cells expressing shRNA against MCP-1 elaborated less than 5% of the MCP-1 level in cells expressing nonspecific shRNA (control cells), and intrapleural delivery of these cells resulted in less MPE (mean MPE volume = 86 and 585 muL, respectively; difference = 499 muL; 95% confidence interval [CI] = 331 to 669 muL; P < .001), reduced MCP-1 levels in the pleural fluid, and lower mortality than when control cells were delivered. Overexpression of MCP-1 in intrapleurally injected B16 melanoma cells led to increased MPE and reduced survival. In mice with MPE, MCP-1 was a potent inducer of vascular permeability, mononuclear recruitment, and, in pleural tumors, of angiogenesis. CONCLUSION: MCP-1 produced by tumor cells is an important determinant of their capacity to induce the formation of MPE and may be a useful target for the treatment of malignant pleural disease.


Subject(s)
Chemokine CCL2/biosynthesis , Neoplasms, Experimental/immunology , Pleural Effusion, Malignant/immunology , Animals , Capillary Permeability , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Disease Models, Animal , Female , Macrophages/immunology , Macrophages/pathology , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Plasmids/genetics , Pleural Effusion, Malignant/genetics , Pleural Effusion, Malignant/pathology , RNA, Small Interfering/genetics , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...